Manual Drainage of the Zebrafish Embryonic Brain Ventricles Please share

advertisement
Manual Drainage of the Zebrafish Embryonic Brain
Ventricles
The MIT Faculty has made this article openly available. Please share
how this access benefits you. Your story matters.
Citation
Chang, Jessica T., and Hazel Sive. “Manual Drainage of the
Zebrafish Embryonic Brain Ventricles.” JoVE no. 70 (2012).
As Published
http://dx.doi.org/10.3791/4243
Publisher
MyJoVE Corporation
Version
Final published version
Accessed
Thu May 26 15:30:52 EDT 2016
Citable Link
http://hdl.handle.net/1721.1/89632
Terms of Use
Article is made available in accordance with the publisher's policy
and may be subject to US copyright law. Please refer to the
publisher's site for terms of use.
Detailed Terms
Journal of Visualized Experiments
www.jove.com
Video Article
Manual Drainage of the Zebrafish Embryonic Brain Ventricles
1
Jessica T. Chang , Hazel Sive
1
1
Department of Biology, Whitehead Institute of Biomedical Research, Massachusetts Institute of Technology
Correspondence to: Hazel Sive at sive@wi.mit.edu
URL: http://www.jove.com/video/4243
DOI: doi:10.3791/4243
Keywords: Neuroscience, Issue 70, Developmental Biology, Medicine, Anatomy, Physiology, Zebrafish, Danio rerio, eCSF, neuroepithelium, brain
ventricular system, brain, microsurgery, animal model
Date Published: 12/16/2012
Citation: Chang, J.T., Sive, H. Manual Drainage of the Zebrafish Embryonic Brain Ventricles. J. Vis. Exp. (70), e4243, doi:10.3791/4243 (2012).
Abstract
Cerebrospinal fluid (CSF) is a protein rich fluid contained within the brain ventricles. It is present during early vertebrate embryonic development
1,2
and persists throughout life. Adult CSF is thought to cushion the brain, remove waste, and carry secreted molecules . In the adult and older
embryo, the majority of CSF is made by the choroid plexus, a series of highly vascularized secretory regions located adjacent to the brain
3-5
6
ventricles . In zebrafish, the choroid plexus is fully formed at 144 hours post fertilization (hpf) . Prior to this, in both zebrafish and other
vertebrate embryos including mouse, a significant amount of embryonic CSF (eCSF) is present . These data and studies in chick suggest that
7
the neuroepithelium is secretory early in development and may be the major source of eCSF prior to choroid plexus development .
8,9
eCSF contains about three times more protein than adult CSF, suggesting that it may have an important role during development . Studies
in chick and mouse demonstrate that secreted factors in the eCSF, fluid pressure, or a combination of these, are important for neurogenesis,
10-20
gene expression, cell proliferation, and cell survival in the neuroepithelium
. Proteomic analyses of human, rat, mouse, and chick eCSF
have identified many proteins that may be necessary for CSF function. These include extracellular matrix components, apolipoproteins, osmotic
21-24
pressure regulating proteins, and proteins involved in cell death and proliferation
. However, the complex functions of the eCSF are largely
unknown.
We have developed a method for removing eCSF from zebrafish brain ventricles, thus allowing for identification of eCSF components and
for analysis of the eCSF requirement during development. Although more eCSF can be collected from other vertebrate systems with larger
embryos, eCSF can be collected from the earliest stages of zebrafish development, and under genetic or environmental conditions that lead to
abnormal brain ventricle volume or morphology. Removal and collection of eCSF allows for mass spectrometric analysis, investigation of eCSF
function, and reintroduction of select factors into the ventricles to assay their function. Thus the accessibility of the early zebrafish embryo allows
for detailed analysis of eCSF function during development.
Video Link
The video component of this article can be found at http://www.jove.com/video/4243/
Protocol
1. Preparing Microinjection Needles and Cell Tram
1.
2.
3.
4.
5.
Fill Eppendorf CellTram oil microinjector apparatus with mineral oil according to manufacturer's instructions.
Prepare microinjection needles by pulling capillary tubes using Sutter instruments needle puller.
Mount needle on micromanipulator connected to Eppendorf CellTram.
Carefully break the tip of the needle. For uniform tip size, measure with a micrometer, or compare to a reference needle.
Fill the length of the needle with oil, using the CellTram to push oil down the length, being careful to avoid any bubbles.
2. Preparing the Embryos
1. Poke holes in a 1% agarose coated dish with a 1-200 μl pipette tip, and remove agarose plugs. Fill dish with embryo media (made according
25
to Westerfield ).
2. Using forceps, dechorionate embryos under stereomicroscope.
3. Transfer dechorionated embryos into agarose coated dish.
25
4. To anesthetize embryos, add Tricaine (0.1 mg/ml) to agarose dish until embryos stop moving (made according to Westerfield ).
Copyright © 2012 Journal of Visualized Experiments
December 2012 | 70 | e4243 | Page 1 of 5
Journal of Visualized Experiments
www.jove.com
3. Draining the eCSF
1. Orient the embryos with their tails in the holes of the agarose and their posterior sides closest to the micromanipulator, allowing for
visualization of the dorsal side of the brain.
2. Position the needle at the rhombomere 0/1 hinge-point or the widest point of hindbrain ventricle (Figure 1A).
3. Carefully pierce roof plate of hindbrain ventricle being sure not to go through the depth of the brain into the yolk.
4. Drain the eCSF using the CellTram and collect fluid in microinjection needle. Be careful to avoid any cells.
4. Collecting the eCSF for Composition Analysis
1.
2.
3.
4.
Once the eCSF is collected in the needle, carefully stop suction from the CellTram.
Move the dish out from under the needle and carefully position the needle into a tube containing the desired buffer.
Using the CellTram, drain the eCSF out of the needle into the buffer trying to avoid contaminating the solution with oil.
In order to remove cells that you have aspirated along with the eCSF, spin eCSF at 10,000 x g. Collect uncontaminated eCSF (supernatant)
and store at -80 °C until ready for further analysis.
5. Reintroduction of Selected Factors
1. Drain the eCSF every 2 hr during desired time interval, since the fluid is replenished within that time period. Between drainage time-points,
remove the needle and store the embryos at 28.5 °C or desired temperature.
2. Load second micropipette needle with test factor.
3. Place needle into micromanipulator attached to microinjector.
4. Adjust injection volume to 1 nl by measuring the drop size in oil and adjusting injection time and pressure.
26
5. Inject 1-2 nl into brain ventricles as described previously .
Representative Results
An example of a drained brain ventricle is shown in Figure 1B-C. Brain ventricles are collapsed as they lack eCSF (Figure 1B vs. C). As seen
in dorsal images (Figure 1B-C, and Figure 2A-D) the hindbrain neuroepithelium does retain its characteristic morphology and seems to be
open despite lack of eCSF likely due to robust hinge-points. However, lateral views (Figure 2A'-D') demonstrate that the hindbrain ventricle
has been drained, consistent with the presence of a thin flexible roof plate epithelium which collapses ventrally. In wild type embryos, eCSF is
continuously produced and refills the brain ventricles 2-3 hr post draining (Figure 2). Therefore, CSF needs to be continually depleted over the
time of interest.
While removal of the fluid at 24 hpf does not disrupt gross morphology or development, removal of eCSF over a longer period of development
may have adverse effects to whole body development. This should be taken into consideration when determining the efficacy of factors injected
into the brain ventricles. Further, to determine whether developmental defects are a result of loss of eCSF or due to a needle puncturing the
brain, an important control is insertion of a needle into the brain without removing eCSF.
We demonstrated that eCSF has a different protein profile than whole embryo extract, and analysis on a SDS-PAGE protein gel demonstrates
that a detectable amount of protein can be collected from zebrafish eCSF (Figure 3). This demonstrates that neuroepithelial cellular
contaminants are at lower levels than eCSF specific proteins, and further that a substantial amount of protein can be collected that is sufficient
for mass spectrometry analysis.
Copyright © 2012 Journal of Visualized Experiments
December 2012 | 70 | e4243 | Page 2 of 5
Journal of Visualized Experiments
www.jove.com
Figure 1. Manually drained brain ventricles. (A) Experimental design. Lateral (left) and dorsal (right) views of where to insert microinjection
needle. Needle (black triangle) is inserted into the roof plate of the hindbrain ventricle at the widest point of the hindbrain. The needle is then
further inserted into the midbrain and forebrain ventricles as designated by the red arrows. (B-C) Examples of 24 hpf manually drained embryo.
The same embryo imaged before draining (B) or after drain (C). (B'-C') Tracings of B-C. Grey line indicates morphology present but not visible. F
= forebrain, M = midbrain, H = hindbrain. Scale bars = 50 μm.
Figure 2. eCSF refills the brain ventricles over time. (A) 24 hpf embryo before draining (B) immediately after t=0 (C) 15, (D) 60, (E) 120, (F)
180, (G) 240, and (H) 300 min (m) after draining. (A-H) Dorsal and (A'-H') lateral brightfield images with anterior to the left. F = forebrain, M =
midbrain, H = hindbrain. Scale bars = 50 μm.
Copyright © 2012 Journal of Visualized Experiments
December 2012 | 70 | e4243 | Page 3 of 5
Journal of Visualized Experiments
www.jove.com
Figure 3. Protein profile differs between 24 hpf whole zebrafish embryo (WE) extract (0.5 μg total protein) and 24 hpf eCSF from 50 embryos.
SDS loading buffer added to collected eCSF to denature proteins, sample run on an 8% Tris-HCl PAGE gel, and detected with Sypro Ruby. *
indicate bands unique to eCSF.
Discussion
Use of this technique to manually drain eCSF from zebrafish brain ventricles will be useful for determining the requirement for eCSF during
development. In addition, this technique will allow description of the eCSF protein profile over the course of embryonic development.
Identification of different proteins during this time will enable further investigation into the function of the CSF and its potential role during
brain development. In amniotes, some factors identified in eCSF (IGF2, FGF2, retinoic acid, and apolipoproteins) have a demonstrated role in
13,17,20,23,27,28
neuroepithelial cell survival, proliferation and neurogenesis
. However, there appear to be hundreds of uncharacterized proteins
in the eCSF, which was obtained after choroid plexus formation, later than the time-points demonstrated here. Additionally, our lab and others
29-31
have identified zebrafish mutants with abnormal brain ventricle size or brain defects
, and may have abnormalities in eCSF composition and
function. This method readily allows for isolating and testing putative abnormal eCSF from mutants or under different environmental conditions.
A powerful use of the zebrafish system is to replace selected factors via injection into the brain ventricles after removal of eCSF, thereby testing
their function during brain development. Small molecules, proteins, and eCSF obtained after genetic or chemical perturbation can be tested.
Reintroduction of eCSF from other species will allow comparison of factors and functions across species and under different pathological
conditions, such as hydrocephalus, complementing and interfacing with mammalian studies. In summary, use of the zebrafish to remove eCSF,
analyze its composition and reintroduce eCSF or specific factors into the brain ventricles, will significantly increase understanding the role and
regulation of eCSF function and that of the brain ventricular system.
Disclosures
No conflicts of interest declared.
Acknowledgements
This work was supported by the National Institute for Mental Health, and National Science Foundation. Special thanks to Dr. Jen Gutzman, Dr.
Amanda Dickinson and other Sive lab members for many useful discussions and constructive criticism, and to Olivier Paugois for expert fish
husbandry.
Copyright © 2012 Journal of Visualized Experiments
December 2012 | 70 | e4243 | Page 4 of 5
Journal of Visualized Experiments
www.jove.com
References
1. Chodobski, A. & Szmydynger-Chodobska, J. Choroid plexus: target for polypeptides and site of their synthesis. Microsc. Res. Tech. 52, 65-82
(2001).
2. Redzic, Z.B., Preston, J.E., Duncan, J.A., Chodobski, A., & Szmydynger-Chodobska, J. The choroid plexus-cerebrospinal fluid system: from
development to aging. Curr. Top. Dev. Biol. 71, 1-52 (2005).
3. Brown, P.D., Davies, S.L., Speake, T., & Millar, I.D. Molecular mechanisms of cerebrospinal fluid production. Neuroscience. 129, 957-970
(2004).
4. Praetorius, J. Water and solute secretion by the choroid plexus. Pflugers Arch. 454, 1-18 (2007).
5. Speake, T., Whitwell, C., Kajita, H., Majid, A., & Brown, P.D. Mechanisms of CSF secretion by the choroid plexus. Microsc. Res. Tech. 52,
49-59 (2001).
6. Garcia-Lecea, M., Kondrychyn, I., Fong, S.H., Ye, Z.R., & Korzh, V. In vivo analysis of choroid plexus morphogenesis in zebrafish. PLoS One.
3, e3090 (2008).
7. Welss, P. Secretory activity of the inner layer of the embryonic mid-brain of the chick, as revealed by tissue culture. The Anatomical Record.
58, 299-302 (1934).
8. Saunders, N.R., Habgood, M.D., & Dziegielewska, K.M. Barrier mechanisms in the brain, II. Immature brain. Clin. Exp. Pharmacol. Physiol.
26, 85-91 (1999).
9. Zheng, W. & Chodobski, A. The blood-cerebrospinal fluid barrier., Taylor and Francis, Boca Raton, Fl, (2005).
10. Salehi, Z. & Mashayekhi, F. The role of cerebrospinal fluid on neural cell survival in the developing chick cerebral cortex: an in vivo study. Eur.
J. Neurol. 13, 760-764 (2006).
11. Parada, C., et al. Embryonic cerebrospinal fluid collaborates with the isthmic organizer to regulate mesencephalic gene expression. J.
Neurosci. Res. 82, 333-345 (2005).
12. Mashayekhi, F. & Salehi, Z. The importance of cerebrospinal fluid on neural cell proliferation in developing chick cerebral cortex. Eur. J.
Neurol. 13, 266-272 (2006).
13. Martin, C., et al. FGF2 plays a key role in embryonic cerebrospinal fluid trophic properties over chick embryo neuroepithelial stem cells. Dev.
Biol. 297, 402-416 (2006).
14. Martin, C., et al. Early embryonic brain development in rats requires the trophic influence of cerebrospinal fluid. Int. J. Dev. Neurosci. 27,
733-740 (2009).
15. Gato, A., et al. Embryonic cerebrospinal fluid regulates neuroepithelial survival, proliferation, and neurogenesis in chick embryos. Anat. Rec.
A Discov. Mol. Cell Evol. Biol. 284, 475-484 (2005).
16. Desmond, M.E., Levitan, M.L., & Haas, A.R. Internal luminal pressure during early chick embryonic brain growth: descriptive and empirical
observations. Anat. Rec. A Discov. Mol. Cell Evol. Biol. 285, 737-747 (2005).
17. Alonso, M.I., Martin, C., Carnicero, E., Bueno, D., & Gato, A. Cerebrospinal fluid control of neurogenesis induced by retinoic acid during early
brain development. Dev. Dyn. 240, 1650-1659 (2011).
18. Miyan, J.A., Zendah, M., Mashayekhi, F., & Owen-Lynch, P.J. Cerebrospinal fluid supports viability and proliferation of cortical cells in vitro,
mirroring in vivo development. Cerebrospinal Fluid Res. 3, 2 (2006).
19. Mashayekhi, F., Bannister, C.M., & Miyan, J.A. Failure in cell proliferation in the germinal epithelium of the HTx rats. Eur. J. Pediatr. Surg. 11
Suppl. 1, S57-59 (2001).
20. Lehtinen, M.K., et al. The cerebrospinal fluid provides a proliferative niche for neural progenitor cells. Neuron. 69, 893-905 (2011).
21. Zappaterra, M.D., et al. A comparative proteomic analysis of human and rat embryonic cerebrospinal fluid. J. Proteome. Res. 6, 3537-3548
(2007).
22. Parvas, M., Parada, C., & Bueno, D. A blood-CSF barrier function controls embryonic CSF protein composition and homeostasis during early
CNS development. Dev. Biol. 321, 51-63 (2008).
23. Parada, C., Gato, A., & Bueno, D. Mammalian embryonic cerebrospinal fluid proteome has greater apolipoprotein and enzyme pattern
complexity than the avian proteome. J. Proteome Res. 4, 2420-2428 (2005).
24. Gato, A., et al. Analysis of cerebro-spinal fluid protein composition in early developmental stages in chick embryos. J. Exp. Zool. A Comp.
Exp. Biol. 301, 280-289 (2004).
25. Westerfield, M., Sprague, J., Doerry, E., Douglas, S., & Grp, Z. The Zebrafish Information Network (ZFIN): a resource for genetic, genomic
and developmental research. Nucleic Acids Res. 29, 87-90 (2001).
26. Gutzman, J.H. & Sive, H. Zebrafish Brain Ventricle Injection. J. Vis. Exp. (26), e1218, doi:10.3791/1218 (2009).
27. Parada, C., Gato, A., & Bueno, D. All-trans retinol and retinol-binding protein from embryonic cerebrospinal fluid exhibit dynamic behaviour
during early central nervous system development. Neuroreport. 19, 945-950 (2008).
28. Parada, C., Escola-Gil, J.C., & Bueno, D. Low-density lipoproteins from embryonic cerebrospinal fluid are required for neural differentiation. J.
Neurosci. Res. 86, 2674-2684 (2008).
29. Kramer-Zucker, A.G., et al. Cilia-driven fluid flow in the zebrafish pronephros, brain and Kupffer's vesicle is required for normal
organogenesis. Development. 132, 1907-1921 (2005).
30. Lowery, L.A. & Sive, H. Initial formation of zebrafish brain ventricles occurs independently of circulation and requires the nagie oko and
snakehead/atp1a1a.1 gene products. Development. 132, 2057-2067 (2005).
31. Lowery, L.A., De Rienzo, G., Gutzman, J.H., & Sive, H. Characterization and classification of zebrafish brain morphology mutants. Anat. Rec.
(Hoboken). 292, 94-106 (2009).
Copyright © 2012 Journal of Visualized Experiments
December 2012 | 70 | e4243 | Page 5 of 5
Download